throbber
FernandoAlbericio
`DepartmentofOrganic
`Chemistry,
`UniversityofBarcelona,
`08028-Barcelona,Spain
`
`OrthogonalProtecting
`GroupsforN a-Aminoand
`C-TerminalCarboxyl
`FunctionsinSolid-Phase
`PeptideSynthesis
`
`a-aminogroupofone
`Abstract:Forthecontrolledsynthesisofeventhesimplestdipeptide,theN
`oftheaminoacidsandtheC-terminalcarboxylgroupoftheothershouldbothbeblockedwith
`suitableprotectinggroups.Formationofthedesiredamidebondcannowoccuruponactivationof
`thefreecarboxylgroup.Aftercoupling,peptidesynthesiscanbecontinuedbyremovalofeitherof
`a-aminogroupofanother
`thetwoprotectinggroupsandcouplingwiththefreeC-terminusorN
`protectedaminoacid.Whenthreefunctionalaminoacidsarepresentinthesequence,thesidechain
`oftheseresiduesalsohastobeprotected.Itisimportantthatthereis ahighdegreeofcompatibility
`betweenthedifferenttypesofprotectinggroupssuchthatonetypemayberemovedselectivelyin
`thepresenceoftheothers.Attheendofthesynthesis,theprotectinggroupsmustberemovedtogive
`thedesiredpeptide.Thus,itisclearthattheprotectionschemeadoptedisoftheutmostimportance
`andmakesthedifferencebetweensuccessandfailurein agivensynthesis.SinceR.B.Merrifield
`introducedthesolid-phasestrategyforthesynthesisofpeptides,thisprerequisitehasbeenreadily
`accepted.Thisstrategyisusuallycarriedoutusingtwomainprotectionschemes:thetert-
`tert-butylmethods.However,forthe
`butoxycarbonyl/benzylandthe9-flourenylmethoxycarbonyl/
`solid-phasepreparationofcomplexorfragilepeptides,aswellasfortheconstructionoflibraries
`ofpeptidesorsmallmoleculesusing acombinatorialapproach, arangeofotherprotectinggroups
`a-aminoand
`isalsoneeded.ThisreviewsummarizesotherprotectinggroupsforboththeN
`C-terminalcarboxylfunctions.©2000JohnWiley &Sons,Inc.Biopoly55:123–139,2000
`
`Keywords:chemicallibraries;combinatorialchemistry;orthogonalstrategy;peptidesynthesis;
`protectinggroup
`
`INTRODUCTION
`
`Peptidesynthesisisbasedontheappropriatecombi-
`nationofprotectinggroupstogetherwithanefficient
`methodfortheactivationofthecarboxylgroupprior
`1 Inthesolid-
`toreactionwiththeaminocomponent.
`phasestrategydevelopedbyMerrifield,theC-termi-
`nalprotectinggroupisinfact apolymericcarrierand,
`consequently,thesynthesisiscarriedoutonaninsol-
`ublesupport. 2 Thesolid-phasemethodnowdomi-
`
`natessyntheticpeptidechemistryandthemajorityof
`peptidesaremadeusingthistechnique.
`SinceMerrifielddescribedthesolid-phaseap-
`proach,onlytwoprotectionschemeshavebeen
`widelyadopted.Thetert-butoxycarbonyl(Boc)/ben-
`zyl(Bzl)strategydependsongraduatedacidlability.
`Thus,whiletheBocgroupisremovedbytrifluoro-
`aceticacid(TFA),Bzl,andrelatedprotectinggroups
`areremovedattheendofthesyntheticstrategywith
`strongacidssuchasHFortrifluoromethanesulfonic.
`
`Biopolymers(PeptideScience),Vol.55,123–139(2000)
`©2000JohnWiley &Sons,Inc.
`
`2
`
`123
`
`MPI EXHIBIT 1006 PAGE 1
`
`MPI EXHIBIT 1006 PAGE 1
`
`

`

`124
`
`Albericio
`
`FIGURE 1 Structure of acid-labile protecting groups.
`
`The main drawback associated with this strategy is a
`lack of flexibility. Long exposure of the peptide chain
`to TFA in the removal the Boc group can also cause
`premature removal of the benzyl protecting group.
`Furthermore, conditions for the removal of Bzl groups
`will always remove the Boc group. Finally, some
`peptides containing fragile sequences will not survive
`the strong acid conditions used to remove the Bzl
`groups. The alternative 9-fluorenylmethoxycarbonyl
`(Fmoc)/tert-butyl (t-Bu) strategy3,4 is based on the
`orthogonal concept5 in the sense that the two protect-
`ing groups belong to independent classes and are
`removed by different mechanisms. The two groups
`can be removed therefore in any order in the presence
`of the group. Orthogonal protection schemes are
`milder because the selective deprotection is governed
`by alternative cleavage mechanisms rather than by
`reaction rates. The Fmoc group6 is removed by pip-
`eridine through a b-elimination reaction and tBu is
`removed by acidolysis with TFA. Although this strat-
`egy has several advantages with respect to the Boc/
`Bzl approach, the conditions are still, in some cases,
`too harsh and can be incompatible with certain se-
`quences. Furthermore,
`the preparation of complex
`molecules such as cyclic or branched systems could
`require the use of other protecting groups.7 In this
`report, other protecting groups for the amino function
`as well as for the carboxyl one are discussed.
`
`Na-AMINO PROTECTING GROUPS
`
`Acid-Labile Protecting Groups
`
`Groups that can be removed through a milder acid in
`comparison to the Boc group are 2-(4-biphenyl)iso-
`(Bpoc),8
`triphenylmethyl
`(trityl,
`propoxycarbonyl
`Trt),9 and a,a-dimethyl-3,5-dimethoxybenzyloxycar-
`bonyl (Ddz)10 (Figure 1).
`
`The Bpoc group can be removed by treatment with
`0.2– 0.5% TFA* in CH2Cl2 and has been used in
`combination with t-Bu side-chain protecting groups in
`the early stage of solid-phase peptide synthesis
`(SPPS).11,12
`The main disadvantage of these derivatives is that
`most are obtained as oils that undergo an autocatalytic
`decomposition to the amino acid, CO2, and the Bpoc
`olefin and its dimer over a half-life of weeks.13 Thus,
`they have to be isolated and stored as cyclohexyl
`ammonium (CHA) or dicyclohexyl
`ammonium
`(DCHA) salts. Liberation of the free carboxyl com-
`pounds is not a straightforward process due to the
`extremely high acid lability of the protecting group.
`Recently, a convenient method for the preparation of
`the O-pentafluorophenyl (O-Pfp) esters has been re-
`ported.13 Most of these compounds are obtained as
`crystalline solids. Furthermore, the Pfp ester is in-
`tended both to protect the highly acid-labile urethane
`from autocatalytic cleavage by the free acid during
`storage and to activate the derivative for acylation
`during peptide coupling reactions. The preparation of
`the Pfp esters of Bpoc-amino acids was performed by
`first reacting the starting amino acid derivative with
`either Bpoc-phenyl carbonate, Bpoc-azide, or Bpoc-
`p-methoxycarbonylphenyl carbonate to form the free
`Bpoc-amino acids, which were immediately esterified
`with dicyclohexylcarbodiimide (DCC).
`SPS of several model peptides has been performed
`using Pfp esters of Bpoc-amino acids (4 equiv) in the
`presence of 1-hydroxybenzotriazole (HOBt; 4 equiv),
`an additive to form in situ the OBt esters and therefore
`to increase the reactivity, and diisopropylethylamine
`(DIEA, 8 equiv) to neutralize Pfp-OH and HOBt, both
`of which are sufficiently acidic to present a threat to
`the stability of the Bpoc group,
`in CH2Cl2/N,N-
`
`* When oxymethyl-containing resins such as poly(ethylene gly-
`col)-based resins are used, higher contents of TFA for this and other
`acid-labile protecting groups could be necessary because some of
`the TFA will be employed for the protonation of the oxymethyl
`moieties.
`
`MPI EXHIBIT 1006 PAGE 2
`
`MPI EXHIBIT 1006 PAGE 2
`
`

`

`Orthogonal Protecting Groups
`
`125
`
`FIGURE 2 Scheme for the alkylation of amide bonds. Adapted from Ref. 22.
`
`dimethylformamide (DMF) (1:1). Removal of the
`protecting group was achieved with 0.5% TFA in
`CH2Cl2 (1 3 3 min 1 1 3 15 min). In this regard, the
`use of scavengers such as benzyl mercaptan and thio-
`phenol has been also reported.14
`The Trt group can be removed in solution with 1%
`15 and 0.1M HOBt in trifluoroethanol
`TFA in CH2Cl2
`(TFE).16 To extend the application of Trt-amino acids
`in solid-phase synthesis, as well as their compatibility
`with hyperacid-labile resins and acid-sensitive bi-
`omolecules, the use of mild acid conditions was in-
`vestigated.17 The results indicate that 0.1M 7-aza-1-
`hydroxybenzotriazole (HOAt)/0.12M Me3SiCl in TFE,
`0.25M Me3SiCl in TFE, 0.2% TFA/1% H2O in CH2Cl2,
`and 3% trichloroacetic acid (TCA) in CH2Cl2 all quan-
`titatively remove the Trt group and are compatible with
`the 3-(4-hydroxymethylphenoxy)propionic acid (AB)
`linker. Furthermore, solutions of 0.2% TFA/1% H2O
`in CH2Cl2, and 3% TCA in CH2Cl2 are compatible
`with the most labile Riniker handle and the synthesis
`of oligonucleotide–peptide conjugates, respectively.
`The synthesis of Trt-amino acids can be carried out
`starting either from the amino acid or the methyl
`ester.9,18 The former method involves in situ trimeth-
`ylsilyl ester formation to protect the carboxylic acid
`followed by reaction with Trt-Cl. The latter method
`requires reaction with Trt-Cl followed by hydrolysis
`of the methyl ester with LiOH/H2O/CH3OH at 25–
`40°C. In our laboratories the second strategy is pre-
`ferred since the overall yields are better (.75% as
`opposed to 40 – 60%) and column chromatography is
`not required to obtain pure derivatives.19
`The main drawback associated with Trt-amino ac-
`ids is that these compounds couple with other amino
`acid derivatives in lower yields when compared with
`carbamate-based protected amino acids.9 SPPS with
`Trt-amino acids have proceeded in good yields
`when N-[(dimethylamino)-1H-1,2,3-triazolo[4,5]b]-
`pyridino-1-ylmethylene]-N-methylmethanaminium
`hexafluorophosphate N-oxide (HATU) in the presence
`of DIEA was used as the coupling reagent. Thus,
`several model peptides have been synthesized using
`
`single couplings with HATU/DIEA (4 equiv:8 equiv)
`for 20 min and with 3% TCA in CH2Cl2 (2 3 3 min)
`for removal of the Trt group.19
`The Trt group could be used in combination with
`an Fmoc strategy in order to avoid diketopiperazine
`(DKP) formation in a similar manner to that described
`earlier for a Boc strategy.20 This procedure avoids the
`presence of the free amino function of the penultimate
`residue and incorporates the third residue with neu-
`tralization in situ. Thus, in those sequences that are
`prone to DKP formation, the following experimental
`protocol should be used: (i) incorporation of the pen-
`ultimate residue as its Trt derivative, (ii9) selective
`detritylation with TFA/H2O/CH2Cl2 (2:1:97) (5 3 1
`min) for AB-type resins or (ii0) TFA/H2O/CH2Cl2
`(0.2:1:99; 5 3 1 min) for Riniker-based resin, and (iii)
`incorporation of the third residue as its Fmoc deriva-
`tive under in situ neutralization/coupling conditions
`mediated by 7-azabenzotriazol-1-yl-N-oxy-tris(pyrro-
`lidino)phosphonium hexafluorophosphate (PyAOP)/
`DIEA (5 equiv each) in DMF for 1 h.21
`The Trt protecting group has also been used in a
`strategy developed to successively alkylate each
`amide bond following its formation.22 Thus, after the
`coupling of an Fmoc-amino acid, the Fmoc group is
`removed and the N-terminal amino function is repro-
`tected with Trt by reaction with Trt-Cl in the presence
`of DIEA. Treatment of the Trt-peptide resin with
`lithium t-butoxide in tetrahydrofuran (THF) leads to
`the formation of the amide anion. Following removal
`of excess base, the alkylating reagent in DMSO is
`reacted with the Trt-peptide resin. The alkylation re-
`action mixture is then removed and the base and
`alkylation treatments repeated to drive the alkylation
`to completion. Removal of the Trt group, followed by
`coupling of the next Fmoc-amino acid allows the
`formation of the next amide bond, which can again be
`alkylated by following the same protocol involving
`the concourse of Trt group (Figure 2).
`The Ddz-amino acids, which are the most commer-
`cially available, are more stable in the presence of
`acids than both the Bpoc and the Trt analogues. The
`
`MPI EXHIBIT 1006 PAGE 3
`
`MPI EXHIBIT 1006 PAGE 3
`
`

`

`126
`
`Albericio
`
`FIGURE 3 Structure of protecting groups that are base labile through a a b-elimination reaction.
`
`Ddz group can be removed with 1–5% TFA in
`CH2Cl2 and the protected amino acids are described
`as being fully compatible with t-Bu side-chain pro-
`tecting groups.23 Furthermore, Ddz is also removed
`by photolysis at wavelengths above 280 nm. This
`property also makes the Ddz group potentially very
`useful in SPS library screening procedures. Ddz de-
`rivatives have a very characteristic uv absorption pat-
`tern with maxima at 230, 276, and 282 nm. Therefore,
`the Ddz fission product allows a very precise deter-
`mination of the initial loading of the first amino acid
`on the resin as well as of the growing Ddz-peptide
`resin.
`Ddz-amino acids have also been used to avoid
`DKP formation when a Backbone Amide Linker
`(BAL) strategy is used.24 As mentioned above for
`AB- and Riniker-type resins, the incorporation of the
`penultimate residue is performed using the Ddz de-
`rivative. Removal of the protecting group is achieved
`with TFA–H2O–CH2Cl2 (3:1:96) for 6 min before
`incorporation of the third residue as its Fmoc deriva-
`tive under in situ neutralization/coupling conditions
`mediated by PyAOP. The advantage of the Ddz pro-
`tecting group over the Trt group is that the former
`couples more efficiently, which is an especially im-
`portant factor when the penultimate residue has to be
`incorporated to a second and/or hindered amine.24
`
`Base-Labile Protecting Groups
`
`Although the use of Fmoc-based SPPS has increased
`enormously in recent years to the point where it is
`now probably the method of choice for the chemical
`synthesis of peptides, there could also be a need to
`search for other, base-labile amino protecting groups.
`The main drawbacks associated with the use of Fmoc-
`amino acids are related to their hydrophobicity. For
`example, some derivatives possess low solubility in
`the solvents commonly used in SPPS, which can
`cause low coupling yields and/or problems associated
`with automation. Solubility issues are more critical
`during the manipulation of Fmoc-protected peptides
`in a convergent strategy.25,26
`
`During the last few years, several Na-amino pro-
`tecting groups have been reported to overcome some
`of the problems described above. Examples of these
`are
`2-[4-(methylsulphonyl)phenylsulphonyl]ethyloxy-
`carbonyl (Mpc),27 2,2-bis-(49-nitrophenyl)ethyloxycar-
`bonyl (Bnpeoc),28 and 2-(2,4-dinitrophenyl)ethyloxycar-
`bonyl (Dnpeoc)29 (See Figure 3).
`Particularly appealing is the 2-(4-nitrophenylsulfo-
`nyl)ethoxycarbonyl (Nsc) group,27,30,31 which can be
`considered from the point of chemical composition to
`be a combination of the Dnpeoc and the Mpc groups,
`since both nitro and sulfonyl groups are incorporated
`into the structure. These two functional groups should
`enhance the solubility of an amino acid and prevent
`hydrophobic interactions between the peptide chains,
`a situation that should subsequently reduce the degree
`of failure or truncated sequences during chain elon-
`gation.32,33 Nsc-amino acids are synthesized easily
`from the corresponding succinimidyl carbonate and
`are crystalline compounds, a physical property
`strongly preferred for automated SPPS. Furthermore,
`the mechanism for the removal of the Nsc group is
`similar to that for Fmoc deprotection and is based
`upon a base-catalyzed b-elimination reaction. There-
`fore, the chemistry and instrumentation described for
`the Fmoc strategy can be readily adapted to this
`protecting group.
`The removal of the Nsc group proceeds at a rate
`three to ten times slower than the removal of the Fmoc
`group, and is dependent on the nature of the base and
`solvent (Table I).34 The high stability of Nsc in DMF
`is of particular importance when this group is to be
`left on for long period of time. Thus, the Nsc group is
`more suitable for use in automatic synthesizers where
`amino acid derivatives are stored in solution for rel-
`atively long periods or when protected peptides are
`left to react in a convergent strategy. In this sense,
`when traces of piperidine remain, the removal of
`Fmoc is still much faster.34 Nsc is clearly a better
`protecting group as far as stability is concerned. Con-
`ditions recommended for the removal of Nsc are 20%
`piperidine in DMF, or preferably DMF– dioxane (1:
`1), for 15 min. The addition of the stronger base
`
`MPI EXHIBIT 1006 PAGE 4
`
`MPI EXHIBIT 1006 PAGE 4
`
`

`

`Orthogonal Protecting Groups
`
`127
`
`Table I Comparison Between Fmoc and Nsc Protected Peptidesa
`
`Cleavage rate (t1/2)
`20% Piperidine/DMF
`1% DBU/20% piperidine/DMF
`Decomposition in DMF solution
`1 week
`3 weeks
`Olefin-amine adduct formation
`Polymerization during removal
`uv monitoring range
`
`a Source: Ref. 34.
`
`Fmoc
`
`10–15 s
`—
`
`Nsc
`
`90–110 s
`12–15 s
`
`10%
`40%
`Fast and reversible
`Yes
`302 nm
`
`, 1%
`2%
`Very fast and irreversible
`No
`380 nm
`
`1,8-diazabicyxlo[5.4.0]undec-7-ene (DBU) can com-
`pensate for the slower rate of Na-deprotection.30 In
`this case, caution is required because the use of DBU,
`even at a concentration of 1% (v/v), can cause aspar-
`timide formation in sensitive sequences.35 Other ad-
`vantages that Nsc has over the Fmoc group are that
`the formation of the olefin-amine adduct is irrevers-
`ible and takes place faster than with Fmoc.34 The Nsc
`group absorbs at 380 nm, which allows selective
`online monitoring during SPPS.
`Coupling kinetic studies have indicated that while
`Fmoc-amino acids at times coupled slightly faster
`than Nsc derivatives, both were globally within the
`same range of reactivity.36
`Premature removal of the Na-Fmoc protecting
`group has been observed during the synthesis of
`polyproline-containing peptides.37 This phenomenon
`is presumably due to the secondary amine character of
`the a-amino function of the Pro. Electrospray mass
`spectroscopy analysis of the sequence H–PPPPPPA–
`NH2 prepared with Nsc–Pro–OH showed no evidence
`of Pro insertion (detected as an [M 1 97] peak),
`which would be indicative of premature deprotection
`of a Pro and the subsequent coupling of another
`protected Pro.36
`
`Table II Racemization Studiesa
`
`H–Gly–aa–L-Phe–NH2
`
`The loss of configuration at the C-terminal car-
`boxyl residue is perhaps the most important side re-
`action in peptide synthesis.38 This is often enhanced
`by the use of highly potent coupling reagents, which
`usually convert the carboxyl function to a derivative
`bearing a good leaving group. Such leaving groups
`tend to increase the acidity of the a-proton and favor
`oxazolone formation, both of which lead to racemiza-
`tion.39 This risk is more accentuated when residues
`such as His,40 Cys,41,42 and Ser43 are incorporated
`into a sequence. To compare the racemization poten-
`tial of Nsc- and Fmoc-amino acids, the model tripep-
`tides H–Gly–Xxx–Phe–NH2 (where Xxx 5 His, Cys,
`and Ser)42,43 were manually synthesized by each
`method in the solid phase.36 Preactivation of the car-
`boxyl group was avoided to minimize racemization of
`the susceptible residues. High performance liquid
`chromatography (HPLC) analysis (Table II) revealed
`that for the Nsc-amino acids, racemization was unde-
`tected for Ser and reduced by more than a half for Cys
`and His. This illustrates the value of Na-Nsc Cys and
`His residues since Fmoc-amino acids gave unaccept-
`able results. Presumably, the higher stability of the
`Nsc-derivatives is correlated with the reduced acidity
`of the b-proton of the protecting group, which trans-
`
`aa: His
`
`aa: Cys
`
`aa: Ser
`
`% L,L
`
`97.5
`2.1
`98.9
`
`% D,L
`
`2.5
`97.9
`1.1
`
`% L,L
`
`89.9
`4.9
`95.7
`
`% D,L
`
`10.1
`95.1
`4.3
`
`% L,L
`
`98.9
`1.0
`99.9
`
`% D,L
`
`1.1
`99.0
`0.1
`
`Fmoc–L-aa–OH
`Fmoc–D-aa–OH
`Nsc–L-aa–OH
`
`a Source: Ref. 36.
`
`MPI EXHIBIT 1006 PAGE 5
`
`MPI EXHIBIT 1006 PAGE 5
`
`

`

`128
`
`Albericio
`
`FIGURE 4 Structure of Dde and ivDde protecting groups and the indazole derivative formed
`during the deprotection step.
`
`lates to a lower acidity of the a-proton of the amino
`acid. However, dedicated protocols for the automatic
`synthesis of peptides with a preactivation step using
`either Nsc or Fmoc chemistry should be developed.
`The use of weaker bases such as collidine (pKa 7.43)
`instead of DIEA (pKa 10.1) is a potential strategy as
`illustrated by studies performed on model sys-
`tems.42– 45
`In conclusion, the Nsc group can be considered an
`alternative to Fmoc for Na-protection.34,36,46 Nsc-
`amino acids are more suitable for use in automatic
`synthesizers where amino acid derivatives are stored
`in solution over relatively long periods or when pro-
`tected peptides are left to react in a convergent strat-
`egy. In this sense, the presence of an Nsc moiety, due
`to its polarity, should also facilitate the purification of
`protected peptides. Furthermore, the use of Nsc de-
`rivatives of Cys and His reduces the racemization of
`these residues when compared with the corresponding
`Fmoc derivatives. The preparation of proline-rich
`peptides occurs without premature removal of the Nsc
`group.
`Although the 1-(4,4-dimethyl-2,6-dioxocyclohexy-
`lidine)ethyl (Dde)47 group can be used for the Na-pro-
`tection, it has been mainly used for the protection of the
`«-amino function of Lys/Orn.48,49 Deprotection, which
`results in the formation of 3,6,6-trimethyl-4-oxo-4,5,6,7-
`tetrahydro-1H-indazole, is carried out by hydrazine–
`DMF (2:98) for 5 min, and can be monitored by the
`absorption at 300 nm. Dde is almost completely stable to
`conditions used to remove the Fmoc group. However,
`this small loss of Dde can compromise the purity of
`large peptides. Furthermore, Dde has also been reported
`3
`N9 migration from side-
`to undergo intramolecular N
`chain or a-amino group to the v-function of Lys/Orn,50
`resulting in the scrambling of the group within the pep-
`tide chain. In order to avoid both side reactions, the
`1-(4,4-dimethyl-2,6-dioxocyclohexylidine)-3-methylbu-
`tyl (ivDde)51 group has been described. This is removed
`in the same conditions than Dde. (See Figure 4.)
`Recently, a new family of Na-amino protecting
`groups, whose removal is achieved by nucleophilic
`addition to a Michael acceptor, has been de-
`
`scribed.52–54 The key deprotection step in the case of
`amines protected by either the 2-tert-butylsulfonyl-2-
`propenoxycarbonyl (Bspoc),53 the 1,1-dioxobenzo-
`(Bsmoc),52 or
`[b]thiophene-2-ylmethyloxycarbonyl
`the 2-methylsulfonyl-3-phenyl-1-prop-2-enyloxycar-
`bonyl (Mspoc)54 groups involves the addition of a
`nucleophilic reagent to the a,b-unsaturated sulfone
`system with the consequent ejection of the carbamate
`ion. (See Figures 5 and 6) There are several major
`advantages of such a process over the classic b-elim-
`ination process involved in the removal of groups
`such as Fmoc or Nsc: (i) the deblocking event is
`simultaneously a scavenging event,
`thus avoiding
`back alkylation of the b-elimination by-product, e.g.,
`dibenzofulvene52; (ii) lower concentrations of second-
`ary amine (piperidine or morpholine) can be used,
`thus minimizing base-catalyzed side reactions such as
`aspartimide formation52,55; and (iii) the method can
`be applied to the technique of rapid continuous solu-
`tion synthesis.55 In turn, the Mspoc group has some
`advantages with respect to the other two approaches
`in question. It is less sensitive to premature deblock-
`ing than the Bspoc and can be assembled from readily
`available materials without recourse to low-valent
`sulfur intermediates, as required in the case of the
`Bsmoc.54
`Due to the different lability in the presence of
`nucleophiles and the difference in the removal mech-
`anism, fluorenylmethyl (Fm)- and 1,1-dioxobenzo
`[b]thiophene-2-ylmethyl
`(Bsm)-based
`protecting
`groups can be orthogonally removed by the appropri-
`ate choice of deblocking reagent.52,55 Thus, Bsmoc
`can be removed in the presence of an Fm ester by
`treatment with 2% tris(2-aminoethyl)amine (TAEA)
`
`FIGURE 5 Structure of protecting groups that are base
`labile through a Michael addition.
`
`MPI EXHIBIT 1006 PAGE 6
`
`MPI EXHIBIT 1006 PAGE 6
`
`

`

`FIGURE 6 Mechanism for the removal of protecting
`groups that are base labile through a Michael addition.
`Adapted from Ref. 22.
`
`and Fmoc can be removed in the presence of a Bsm ester
`by N-methyl-tert-butylamine, the steric hindrance of
`which avoids extensive Michael-like addition.
`Several model peptides have been synthesized in
`solid-phase using Bsmoc-amino acids.52 The removal
`of the Bsmoc group was accomplished with 2–5%
`piperidine in DMF. The use of 2% piperidine in DMF
`for 7 min leads to a reduction in the extent of aspar-
`timide formation, e.g., 4.8 vs 11.6% obtained when
`20% piperidine is used for the removal of Fmoc group
`in a sequence prone to give this side reaction. An
`additional advantage of Bsmoc over Fmoc assembly
`was noted in the case of pentapeptide H–Tyr–Aib–
`Aib–Phe–Leu–OH, where the difficult Aib–Aib cou-
`pling was favored by about 13%.
`
`Pd-Labile Protecting Groups
`
`In 1950 the allyloxycarbonyl (Alloc) group was in-
`troduced by Stevens and Watanabe for the protection
`of amine and alcohol functions.56 They reported that
`cleavage of the Alloc group was accomplished by
`catalytic hydrogenolysis using platinum or palladium
`catalysts, the use of metallic sodium in liquid ammo-
`nia, or the use of phosphonium iodide in glacial acetic
`acid (HOAc). Unfortunately, these methods for the re-
`moval of the Alloc group were not straightforward,
`which limited its use in organic synthesis. With the
`palladium-catalyzed allyl removal reaction developed
`by Tsuji57 and Trost,58 the potential of the allyl protect-
`ing group was recognized.59,60 The deprotection step
`involves a palladium-catalyzed transfer of the allyl
`unit to various nucleophiles/scavengers in the pres-
`ence of a proton source (Figure 7). Thus, allyl-based
`groups are orthogonal with both base-labile, such as
`Fm, and acid-labile, such as t-Bu, protecting groups.
`
`Orthogonal Protecting Groups
`
`129
`
`Allyl chemistry has mainly been used in solid-
`phase strategies either as a handle or as a side-chain
`protecting strategy. Thus, Kunz and co-workers61 first
`introduced the use of 4-bromocrotonic acid coupled to
`aminomethyl polystyrene resin as an allylic anchor.
`Since then, other handles have also been used for the
`preparation of a broad range of peptides.25,60,62 Hud-
`son and co-workers63 and Loffet and Zhang64 inde-
`pendently investigated the use of the allyl group as a
`side-chain protecting group. The three-dimensional
`orthogonal
`strategy (Fmoc/t-Bu/allyl) has been
`mainly used for the preparation of cyclic peptides.65,66
`The Na-Alloc-amino acids are synthesized easily
`from the corresponding chloroformates67 using the
`method developed by Bolin and co-workers for the
`corresponding Fmoc derivatives.68 It should be men-
`tioned that even for hindered amino acids such as Val
`the formation of the protected dipeptide has been
`observed.69 This method involves the in situ prepara-
`tion of the trimethylsilyl derivatives and subsequent
`reaction with the chloroformate. Although these de-
`rivatives are obtained as oils, they can be crystallized,
`purified, and stored as their dicyclohexylammonium
`salts. Alternatively,
`the pentafluorophenyl esters,
`which are crystalline, can be easily prepared using the
`procedure described by Green and Berman for the
`Fmoc derivatives.70 These active esters represent an
`excellent method of storage for Na-Alloc amino acids
`and as active species can be used directly for synthetic
`purposes.67 In this case, it is convenient to perform
`the coupling in the presence of an equivalent of HOBt
`or HOAt in order to accelerate the process.
`The main concern with the use of the Alloc group
`for Na-amino protection is the formation of al-
`lylamines. The consequences of this side reaction can
`be more important when the Alloc group is used in a
`repetitive way for temporary Na-amino protection in
`SPPS. Allylamine formation can take place during the
`Pd-catalyzed removal of allylcarbamates and may oc-
`cur in different ways.60
`First of all, the free amine may compete with the
`nucleophilic allyl-group scavenger in the trapping of
`the p-allyl palladium complex. Therefore, it is better
`to use deprotection systems that lead to the protonated
`
`FIGURE 7 Palladium-catalyzed deprotection of allyl carbamates and carboxylates.
`
`MPI EXHIBIT 1006 PAGE 7
`
`MPI EXHIBIT 1006 PAGE 7
`
`

`

`130
`
`Albericio
`
`FIGURE 8 Mechanism of allylamine formation when protonic reversible allyl-group scavengers
`are used. Adapted from Ref. 60.
`
`amine or other masked non-nucleophilic forms of the
`amine, or to use the allyl scavenger in large excess.
`With the protonic reversible allyl-group scavengers
`ReH, allylamine may also be formed through an
`equilibration process (Figure 8). Finally, Alloc-
`amines are known to undergo decarboxylative rear-
`rangement to allyl amines in the presence of Pd cat-
`alysts and in the absence of nucleophiles.71
`The choice of an appropriate allyl scavenger is
`clearly a key factor for the establishment of a conve-
`nient strategy for the use of Na-Alloc as a temporary
`protecting group in SPPS. Guibe´ and co-workers72
`described the first SPPS using this protecting group.
`In this work, the removal of the Alloc group was
`effected by the hydrostannolytic procedure, which
`utilizes tributyltin hydride as the allyl group scaven-
`ger. This procedure was selected on the grounds that
`the hydrostannolytic procedure is extremely fast, be-
`ing complete within a few minutes (thus minimizing
`the risk of allylamine formation), and very selective,
`since tributyltin hydride is nonbasic and is, at room
`temperature, inert toward almost any reducible func-
`tions that do not interact with soluble palladium cat-
`alysts. On using this method the peptide was obtained
`in a moderate, nonoptimized yield of 25%. Two draw-
`
`backs, however, are associated with the use of tribu-
`tyltin hydride. The first is that tributyltin hydride is
`not easy to handle and, in particular, shows a propen-
`sity, in the presence of various agents including pal-
`ladium catalysts, to decompose into hexabutyldistan-
`nane and dihydrogen.72 The second drawback is as-
`sociated with the toxicity of tin compounds and the
`difficulty often encountered in their complete removal
`from the desired end products (this last difficulty is
`minimized in the case of synthesis on solid supports).
`Other allyl group scavengers have been investigated
`with the aim of developing alternative tin-free meth-
`ods that are well suited for repetitive Alloc removal
`and compatible with the presence of Fmoc and t-Bu
`protecting groups.67 Table III shows the results of a
`study carried out in which the Alloc derivative of the
`N-methylbenzylamine was taken as a model and
`shows a higher propensity to form allylamine than
`Alloc derivatives of primary amines. The study in-
`cludes the pronucleophilic species (NDMBA) and
`thiosalicyclic acid (TSA) proposed by Kunz73 and
`Geneˆt,74 respectively, and pseudometallic hydrides,
`2), initially proposed by Zhu
`i.e., borohydrides (BH4
`and co-workers,75 phenylsilane (PhSiH3) proposed by
`Guibe´ and co-workers,76 and several as yet untested
`
`Table III Palladium-Catalyzed Deprotection [Pd(Ph3)4] of the
`Alloc Derivative of N-Methylbenzylamine in the Presence of
`Various Allyl Group Scavengersa
`
`Allyl Group Scavenger
`
`Yield (%)
`HON(Me)CH2Ph
`AllON(Me)CH2Ph
`
`2
`
`NDMBA
`TSA
`PhSiH3
`Bu4N1BH4
`z BH3
`NH3
`(Me)2NH z BH3
`z BH3
`t-BuNH2
`(Me)3N z BH3
`Py z BH3
`
`a Source: Ref. 67.
`
`100
`100
`95
`60
`99.6 to 100
`100
`96 to 97
`0
`0
`
`0
`0
`5
`40
`0 to 0.4
`0
`3 to 4
`100
`100
`
`MPI EXHIBIT 1006 PAGE 8
`
`MPI EXHIBIT 1006 PAGE 8
`
`

`

`Orthogonal Protecting Groups
`
`131
`
`FIGURE 9 Palladium-catalyzed deprotection of allyl carbamates in the presence of boranes.
`Adapted from Ref. 67.
`
`z BH3,
`including NH3
`z BH3, (Me)3NH z BH3, and
`
`amine-borane complexes
`(Me)2NH z BH3, t-BuNH2
`Py z BH3.
`The pronucleophilic scavengers NDMBA and
`TSA, and the amine borane complexes H3N z BH3 and
`Me2NH z BH3, were found to lead selectively to
`N-methylbenzylamine. Meanwhile, concurrent and in-
`creasing formation of N-allyl- and N-methylben-
`z BH3
`zylamine was observed in the series t-BuNH2
`2. As far as the experiments with Py z
`, BH4
`# PhSiH3
`BH3 and Me3N z BH3 are concerned, these reactions
`led exclusively to the N-allylated amine. Clearly the
`tertiary amine– borane complexes do not act as allyl
`group scavengers at all. From the above experiments
`and with regard to selectivity, the pronucleophilic
`species NDMBA and TSA, on the one hand, and the
`amine– borane complexes H3N z BH3 and Me2NH z
`BH3, on the other hand, appear to be the best-suited
`scavengers for the palladium-catalyzed removal of
`N-Alloc groups. With regard to the reactivity, how-
`ever, the amine– borane complexes that lead to com-
`plete deprotection in less than 10 min are far superior
`to the pronucleophilic species, for which reaction
`times of between 30 and 90 min are required for the
`reaction to reach completion. Therefore, only the two
`amine– borane complexes H3N z BH3 and Me2NH z
`BH3 can be recommended as deprotecting agents for
`use in SPPS strategies. It should be noted that, in a
`similar way to that observed with other pseudometal-
`lic hydrides such as tributyltin hydride72 or phenylsi-
`lane,76,77 the deprotection of N-Alloc derivatives with
`amine– borane complexes does not lead to the free
`amines but to the corresponding boron carbamates,
`which are readily hydrolyzed and decarboxylated
`upon exposure, for instance, to small amounts of
`water or silica. (See Figure 9.)
`The stability of the Fmoc group and the ease of
`reduction of the Trp residue were assessed against
`these amine– borane complexes. It was found that
`Ac–Trp–OMe and Fmoc–Trp(Boc)–OMe were left
`unchanged after exposure at 25°C for 48 h to H3N z
`BH3 and Me2NH z BH3 either in the absence or the
`presence of Pd(Ph3)4. On the other hand, even in the
`absence of catalyst, Boc–Trp(CHO)–OMe is slowly

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket